Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hideki Hirabayashi is active.

Publication


Featured researches published by Hideki Hirabayashi.


Drug Metabolism and Disposition | 2011

Quantitative Investigation of the Impact of P-Glycoprotein Inhibition on Drug Transport across Blood-Brain Barrier in Rats

Hiroshi Sugimoto; Hideki Hirabayashi; Yoshiaki Kimura; Atsutoshi Furuta; Nobuyuki Amano; Toshiya Moriwaki

The magnitude of P-glycoprotein [(P-gp)/multidrug resistance protein 1 (MDR1)]-mediated drug-drug interaction (DDI) at the blood-brain barrier (BBB) in rats was estimated by in vitro-in vivo correlation (IVIVC). In in vitro studies, rat Mdr1a-expressing LLC-PK1 cells were examined for the evaluation of P-gp inhibitory activity using digoxin as a P-gp probe substrate. The in vitro Ki value was calculated using a modified corrected flux ratio that reflects the P-gp function. In in vivo studies, digoxin with or without P-gp inhibitors was administered to rats by constant intravenous infusion to evaluate the effect of P-gp inhibition on digoxin transport to the brain under steady-state conditions. In the presence of elacridar, the brain-to-plasma concentration ratio (Kp,brain) of digoxin was approximately 14 times the control value. However, no significant change in the Kp,brain was observed in the presence of clinically used P-gp inhibitors, with the exception of cyclosporine A. A positive correlation was found between the in vivo Kp,brain of digoxin and [I,unbound/Ki] (where I,unbound is the unbound plasma concentration of P-gp inhibitors). Compounds with [I,unbound/Ki] values of >1 increased Kp,brain of digoxin in rats. In summary, we used a quantitative approach to evaluate the impact of P-gp-mediated DDI at the rat BBB. We successfully established the IVIVC, which indicated the potential DDI in the presence of potent P-gp inhibitors. On the basis of the IVIVC in rats and Ki values in human MDR1, we speculated that clinically used P-gp inhibitors do not cause DDI at the human BBB, because none of the compounds studied showed [I,unbound/Ki] values of >1 at therapeutic doses.


Journal of Pharmaceutical Sciences | 2011

Establishment of In Vitro P‐Glycoprotein inhibition assay and its exclusion criteria to assess the risk of drug–drug interaction at the drug discovery stage

Hiroshi Sugimoto; Shin-ichi Matsumoto; Miho Tachibana; Shin-Ichi Niwa; Hideki Hirabayashi; Nobuyuki Amano; Toshiya Moriwaki

The decision tree to determine whether the P-glycoprotein (P-gp)/multidrug resistance protein 1 (MDR1)-mediated drug-drug interaction (DDI) study is recommended has been proposed by the International Transporter Consortium. We, therefore, designed an in vitro P-gp inhibition assay and determined the appropriate risk criteria for P-gp-mediated DDI at the drug discovery stage. Effects of P-gp inhibitors on digoxin transport across a monolayer of MDR1-expressing cells were examined. The IC(50) (half-maximal inhibitory concentration) values generated from the efflux ratio (ER) were smaller than those generated from basolateral-to-apical directional apparent permeability. The difference in IC(50) values was kinetically described in a compartment model analysis. This analysis indicated that ER is a highly sensitive parameter that can be used for the degree of P-gp inhibition. Considering IC(50) values and the increase in digoxin exposure in clinical DDI studies, the risk criteria of [I(2)]/IC(50) = 30 ([I(2)], theoretically maximal gastrointestinal concentration) was the optimal cutoff value to predict a clinically relevant DDI. We also investigated whether the IC(50) value itself is applicable to assess the DDI risk. In conclusion, compounds with IC(50) values less than 2 μM exhibit high risk for P-gp-mediated DDIs. However, compounds with IC(50) values greater than or equal to 2 μM are inconclusive because clinical doses should be considered for the precise DDI risk assessment.


Journal of Chromatography B | 2011

Development of a quantification method for digoxin, a typical P-glycoprotein probe in clinical and non-clinical studies, using high performance liquid chromatography-tandem mass spectrometry: the usefulness of negative ionization mode to avoid competitive adduct-ion formation.

Hideki Hirabayashi; Hiroshi Sugimoto; Shin-ichi Matsumoto; Nobuyuki Amano; Toshiya Moriwaki

Highly sensitive and accurate liquid chromatography-tandem mass spectrometry (LC/MS/MS) methods have been developed and validated for measuring digoxin (DGX), a typical P-glycoprotein probe, in human plasma, rat plasma, and rat brain. We extracted DGX and deuterium-labeled DGX (as internal standard) from sample fluids under basic conditions using acetonitrile and sodium chloride-saturated 0.1 mol/L sodium hydroxide. The upper organic layer was diluted with distilled water, and the resulting solution was injected into an LC/MS/MS system in negative ionization mode. Chromatographic separation was achieved on a C(18)-ODS column in the gradient mobile phase, which comprised 0.05% (w/v) ammonium carbonate (pH 9.0) and methanol at a flow rate of 0.7 mL/min. Regardless of the type of biological matrix, intra-day and inter-day validation tests demonstrated good linearity of calibration curves within ranges of 0.1-10 ng/mL for plasma and 0.5-50 ng/g for rat brain and gave excellent accuracy and precision of quality control samples at 4 concentration levels. Unlike existing methods, our approach uses negative ionization to avoid competitive adduct formation of DGX. Our method showed higher sensitivity and wider applicability to various types of biological matrices than existing methods. Our method will support clinical and preclinical investigation of in vivo P-glycoprotein functionality using DGX.


Xenobiotica | 2012

Evaluation of cytochrome P450-mediated drug–drug interactions based on the strategies recommended by regulatory authorities

Yohei Kosugi; Hideki Hirabayashi; Tomoko Igari; Yasushi Fujioka; Yoko Hara; Teruaki Okuda; Toshiya Moriwaki

Herein, we aimed to evaluate the recently proposed risk assessment strategies of a cytochrome P450 (CYP) mediated drug–drug interaction (DDI) according to the European Medicines Evaluation Agency (EMEA) draft guideline, and discuss the differences between this guideline and the Food and Drug Administration (FDA) draft guidance. A retrospective study on reported 35 clinical DDI cases revealed that the EMEA assessment successfully predicts moderate-to-strong DDIs, i.e. drugs that cause more than 2-fold increase in the area under the curve in the presence and absence of CYP inhibitor (AUCi/AUC); however, EMEA tends to overlook weak DDIs with AUCi/AUC ≤ 2 to > 1.25. For CYP3A4 inhibitors, even clinically insignificant DDIs were overemphasized if the intestinal DDI is considered. The differences between unbound fraction in plasma and microsomes account for the discrepancies in DDI risk assessment results between EMEA and FDA assessments. Comparing two assessment results for CYP2D6 and CYP2C9 inhibitors, the FDA assessment suggested potential DDI risks for sulphinpyrazone and amitriptyline, while the EMEA assessment indicated no potential risk for these drugs. Through a retrospective study, we showed practical differences in the DDI assessment strategies of EMEA and FDA and suggested improvements in their current strategies.


Drug Metabolism and Disposition | 2013

Retrospective Analysis of P-Glycoprotein–Mediated Drug-Drug Interactions at the Blood-Brain Barrier in Humans

Hiroshi Sugimoto; Hideki Hirabayashi; Nobuyuki Amano; Toshiya Moriwaki

To date, the in vitro–in vivo correlation (IVIVC) of P-glycoprotein (P-gp)–mediated drug-drug interaction (DDI) at the blood-brain barrier (BBB) in rats indicated that the cutoff value to significantly affect the brain penetration of digoxin was [I,unbound/Ki] of 1, where I,unbound is the unbound plasma concentration of P-gp inhibitors. On the basis of the IVIVC in rats, we speculated that clinically used P-gp inhibitors do not cause DDI at the human BBB, because none of the compounds studied was [I,unbound/Ki]>1 at therapeutic doses. Recently, positron emission tomography studies with P-gp substrates, such as [11C]verapamil, [11C]N-desmethyl loperamide, and [11C]loperamide, together with potent P-gp inhibitors, have indicated that increases in the influx rate constant for brain entry were observed in humans. Therefore, we aimed to retrospectively analyze the results of P-gp–mediated DDIs with in vitro P-gp inhibition assays and to confirm the appropriate cutoff value. In vitro P-gp inhibition assays using verapamil, N-desmethyl loperamide, and loperamide as P-gp probe substrates were performed in human multidrug resistance protein 1-expressing LLC-PK1 cells. The efflux ratios decreased in the presence of P-gp inhibitors, and the Ki of tariquidar was 10 nmol/L, regardless of probe substrates. Taking the in vitro Ki and unbound plasma concentrations in clinical DDI studies together, the criterion [I,unbound/Ki] of 1 was an appropriate cutoff limit to observe significant P-gp–mediated DDI at the BBB in humans. On the other hand, no significant DDI was observed in cases in which [I,unbound/Ki] was less than 0.1. This criterion was comparable to the previous IVIVC result in rats.


Xenobiotica | 2017

In vitro metabolism of TAK-438, vonoprazan fumarate, a novel potassium-competitive acid blocker

Hitomi Yamasaki; Naohiro Kawaguchi; Masami Nonaka; Junzo Takahashi; Akio Morohashi; Hideki Hirabayashi; Toshiya Moriwaki; Satoru Asahi

Abstract 1. TAK-438, vonoprazan fumarate, is a novel orally active potassium-competitive acid blocker, developed as an antisecretory drug. In this study, we investigated the in vitro metabolism of 14C-labeled TAK-438. In human hepatocytes, M-I, M-II, M-III and M-IV-Sul were mainly formed, and these were also detected in clinical studies. N-demethylated TAK-438 was also formed as an in vitro specific metabolite. Furthermore, CYP3A4 mainly contributed to the metabolism of TAK-438 to M-I, M-III, and N-demethylated TAK-438, and CYP2B6, CYP2C19 and CYP2D6 partly catalyzed the metabolism of TAK-438. The sulfate conjugation by SULT2A1 also contributed to the metabolism of TAK-438 to form TAK-438 N-sulfate, and CYP2C9 mediated the formation of M-IV-Sul from TAK-438 N-sulfate. The metabolite M-IV, which could be another possible intermediate in the formation of M-IV-Sul, was not observed as a primary metabolite of TAK-438 in any of the in vitro studies. 2. In conclusion, TAK-438 was primarily metabolized by multiple metabolizing enzymes including CYP3A4, CYP2B6, CYP2C19, CYP2D6, and a non-CYP enzyme SULT2A1, and the influence of the CYP2C19 genotype status on gastric acid suppression post TAK-438 dosing could be small. The multiple metabolic pathways could also minimize the effects of co-administrated CYP inhibitors or inducers on the pharmacokinetics of TAK-438.


Xenobiotica | 2017

Comparison of predictability for human pharmacokinetics parameters among monkeys, rats, and chimeric mice with humanised liver

Maki Miyamoto; Shinji Iwasaki; Ikumi Chisaki; Sayaka Nakagawa; Nobuyuki Amano; Hideki Hirabayashi

Abstract 1. The aim of the present study was to evaluate the usefulness of chimeric mice with humanised liver (PXB mice) for the prediction of clearance (CLt) and volume of distribution at steady state (Vdss), in comparison with monkeys, which have been reported as a reliable model for human pharmacokinetics (PK) prediction, and with rats, as a conventional PK model. 2. CLt and Vdss values in PXB mice, monkeys and rats were determined following intravenous administration of 30 compounds known to be mainly eliminated in humans via the hepatic metabolism by various drug-metabolising enzymes. Using single-species allometric scaling, human CLt and Vdss values were predicted from the three animal models. 3. Predicted CLt values from PXB mice exhibited the highest predictability: 25 for PXB mice, 21 for monkeys and 14 for rats were predicted within a three-fold range of actual values among 30 compounds. For predicted human Vdss values, the number of compounds falling within a three-fold range was 23 for PXB mice, 24 for monkeys, and 16 for rats among 29 compounds. PXB mice indicated a higher predictability for CLt and Vdss values than the other animal models. 4. These results demonstrate the utility of PXB mice in predicting human PK parameters.


ACS Omega | 2017

A Short-Length Peptide YY Analogue with Anorectic Effect in Mice

Naoki Nishizawa; Ayumu Niida; Yasushi Masuda; Satoshi Kumano; Kotaro Yokoyama; Hideki Hirabayashi; Nobuyuki Amano; Tetsuya Ohtaki; Taiji Asami

Peripheral administration of PYY3–36, a fragment of peptide YY (PYY), has been reported to reduce food intake by activating the neuropeptide Y2 receptor (Y2R). An N-terminally truncated PYY analogue, benzoyl-[Ala26,Ile28,31]PYY(25–36) (1), showed a relatively potent agonist activity for Y2R but a weak anorectic activity by intraperitoneal administration (2000 nmol/kg) in lean mice because of its markedly poor biological stability in the mouse serum. Notably, two cyclohexylalanine (Cha) substitutions for Tyr residues at positions 27 and 36 (4) improved the stability in the mouse serum concomitant with enhanced anorectic activity. Further optimization at positions 27, 28, 30, and 31 revealed that 21, containing Cha28 and Aib31 residues, showed a more potent anorectic activity than PYY3–36 at a low dose of 300 nmol/kg. The minimum effective dose by intraperitoneal administration of 21 was 30 nmol/kg (ca. 52 μg/kg) in mice, suggesting the biologic potential of short-length PYY3–36 analogues with a potent anorectic effect.


ACS Medicinal Chemistry Letters | 2017

Antiobesity Effect of a Short-Length Peptide YY Analogue after Continuous Administration in Mice

Naoki Nishizawa; Ayumu Niida; Yasushi Masuda; Satoshi Kumano; Kotaro Yokoyama; Hideki Hirabayashi; Nobuyuki Amano; Tetsuya Ohtaki; Taiji Asami

Gastrointestinal peptides such as peptide YY (PYY) can regulate appetite, which is relevant to the study of obesity. The intraperitoneal bolus administration of PYY3-36 and a 12-amino acid PYY analogue, benzoyl-[Cha27,28,36,Aib31]PYY25-36 (1), showed similar anorectic activity by activating the Y2 receptor (Y2R). However, food intake inhibition and body weight loss were not observed upon continuous subcutaneous administration of 1 with osmotic pumps in diet-induced obese (DIO) mice. N-Terminal elongation of 1, together with amino acid substitution at position 24, led to a hydrophilic 14-amino acid peptide, Ac-[d-Hyp24,Cha27,28,36,Aib31]PYY23-36 (18), that showed higher affinity and more potent agonist activity for Y2R and a robust anorectic activity with potency similar to that of PYY3-36. In addition, the continuous subcutaneous administration of 18 at 0.3 mg/(kg·day) induced significant body weight loss in DIO mice. These results suggest that a short-length PYY analogue can be a lead compound for antiobesity therapy in a sustained-release formulation.


Journal of Medicinal Chemistry | 2017

Potent Body Weight-Lowering Effect of a Neuromedin U Receptor 2-selective PEGylated Peptide

Yoko Kanematsu-Yamaki; Naoki Nishizawa; Tomoko Kaisho; Hiroaki Nagai; Taisuke Mochida; Tomoko Asakawa; Hiroshi Inooka; Katsuko Dote; Hisashi Fujita; Kouta Matsumiya; Hideki Hirabayashi; Junichi Sakamoto; Tetsuya Ohtaki; Shiro Takekawa; Taiji Asami

Neuromedin U (NMU) is a neuropeptide that mediates a variety of physiological functions via its receptors, NMUR1 and NMUR2. Recently, there has been an increased focus on NMU as a promising treatment option for diabetes and obesity. A short form of NMU (NMU-8) has potent agonist activity for both receptors but is metabolically unstable. Therefore, we designed and synthesized NMU-8 analogues modified by polyethylene glycol (PEG; molecular weight, 20 kDa; PEG20k) via a linker. 3-(2-Naphthyl)alanine substitution at position 19 increased NMUR2 selectivity of NMU-8 analogues with retention of high agonist activity. Compound 37, an NMUR2-selective PEG20k analogue containing piperazin-1-ylacetyl linker, exhibited a potent body weight-lowering effect with concomitant inhibition of food intake in a dose-dependent manner (body weight loss of 12.4% at 30 nmol/kg) by once-daily repeated dosing for 2 weeks in mice with diet-induced obesity.

Collaboration


Dive into the Hideki Hirabayashi's collaboration.

Top Co-Authors

Avatar

Nobuyuki Amano

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Toshiya Moriwaki

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Naoki Nishizawa

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Taiji Asami

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Ayumu Niida

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Tetsuya Ohtaki

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Hiroshi Sugimoto

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Kotaro Yokoyama

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Satoshi Kumano

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Shiro Takekawa

Takeda Pharmaceutical Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge